EP1687411B1 - Identification et isolation de cellules souches hematopoietiques - Google Patents

Identification et isolation de cellules souches hematopoietiques Download PDF

Info

Publication number
EP1687411B1
EP1687411B1 EP20040809807 EP04809807A EP1687411B1 EP 1687411 B1 EP1687411 B1 EP 1687411B1 EP 20040809807 EP20040809807 EP 20040809807 EP 04809807 A EP04809807 A EP 04809807A EP 1687411 B1 EP1687411 B1 EP 1687411B1
Authority
EP
European Patent Office
Prior art keywords
cells
hscs
cell sample
bone marrow
hematopoietic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP20040809807
Other languages
German (de)
English (en)
Other versions
EP1687411A4 (fr
EP1687411A2 (fr
Inventor
Omer H. Yilmaz
Mark J. Kiel
Sean Morrison
Toshihide Iwashita
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Michigan
Original Assignee
University of Michigan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Michigan filed Critical University of Michigan
Publication of EP1687411A2 publication Critical patent/EP1687411A2/fr
Publication of EP1687411A4 publication Critical patent/EP1687411A4/fr
Application granted granted Critical
Publication of EP1687411B1 publication Critical patent/EP1687411B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors

Definitions

  • the present invention is defined in the claims and relates to methods of identifying, collecting and isolating hematopoietic stem cells (HSCs) and compositions of purified HSCs. Specifically, the present invention provides methods of isolating and collecting CD150 + HSCs, CD48 - HSCs, and CD244 - HSCs. The present invention also relates to purified cell samples with enriched CD150 + HSCs, CD48 - HSCs, and CD244 - HSCs populations, as well as methods of treating subjects with such compositions.
  • HSCs hematopoietic stem cells
  • HSCs Hematopoietic stem cells in the bone marrow are responsible for the production of mature blood cells throughout adult life. HSCs are identified by their ability to self-renew and undergo multilineage differentiation to form all major lineages of blood cells. HSCs can be transplanted into irradiated subjects, where they will engraft and give rise large numbers of diverse types of blood cells including myeloid, B, and T cells.
  • the state of the art for HSC purification relies on complex combinations of up to 12 markers, which target specific cell surface proteins, to isolate HSCs from bone marrow or other hematopoietic tissues by flow-cytometry.
  • the highest enrichment of HSC activity yet reported describes combinations of markers, such those used to isolate Thy-1.1 lo Sca-1 + lineage - Mac-1 - CD4 - c-kit + cells (hereafter referred to as Mac-1 - CD4 - c-kit + ), from which one out of every five intravenously injected cells are able to home to bone marrow and engraft (see, e.g., Uchida et al.., J Exp Med.
  • HSCs are characterized by the ability to undergo long-term multilineage reconstitution (for extended periods of time and usually for the life of a subject, in the case of a murine subject, for more than 16 weeks to life), whereas other populations of hematopoietic progenitors only transiently form mature blood cells.
  • HSCs give rise to non-self-renewing multipotent progenitors (MPPs) that can be isolated as Thy-1.1 lo Sca-1 + Mac-1 lo CD4 lo bone marrow cells (See, Morrison et al., 1994 and Morrison et al. 1997 supra). These cells give rise to myeloid B and T cells for less than 6 weeks after injection into irradiated mice, for example.
  • MPPs multipotent progenitors
  • Highly purified HSCs are increasingly being used clinically, such as for autologous transplants into patients after high-dose chemotherapy.
  • immune effector cells such as lymphocytes
  • cancer cells Since few markers or cell surface proteins have been identified that are highly specific to HSCs, it is not possible to identify or collect these cells based on simple combinations of one or two markers. As a result, it has only been possible to highly purify HSCs using complex combinations of many markers. These complex combinations of markers have not been practical to use clinically.
  • the present invention is defined in the claims and provides methods of identifying and collecting hematopoietic stem cells (HSCs) and compositions highly enriched for HSCs.
  • HSCs hematopoietic stem cells
  • the present invention provides methods of purifying HSCs (e.g. from a hematopoietic tissue sample) based on the presence or absence of SLAM family member proteins, and especially CD 150, CD48, and CD244.
  • the present invention provides methods of identifying and collecting CD150 + HSCs, CD150 + CD48 - HSCs, CD 150 + CD244 - HSCs, and CD150 + CD48 - CD244 - HSCs from a cell sample, as well as purified samples having an increased level ofCD150 + HSCs, CDI50 + CD48 - HSCs, CD150 + CD244 - HSCs, and CD150 + CD48 - CD244 - HSCs.
  • the present invention also relates to methods of treating subjects with such compositions.
  • the present invention provides methods comprising a) providing a first sample comprising hematopoietic stem cells (HSCs) and b) purifying the first sample under conditions such that a purified sample is generated, wherein the purified sample comprises a higher percent of CD 150 + HSCs, CD48 - HSCs, or CD244 - HSCs than present in the first sample.
  • the present invention provides compositions purified by the above method.
  • the present invention provides methods comprising: purifying cells from a first sample, based on the positive expression of CD150 protein or negative expression of CD48 protein or CD244 protein, in order to generate a purified cell sample, wherein the cells are selected from at least one of the following: CD 150 + cells, CD48 - cells, and CD244 - cells, and wherein the purified cell sample comprises a higher percent of HSCs than are present in the first sample.
  • the present invention provides compositions purified by the above method.
  • the present invention relates to methods comprising; a) providing: i) a first cell sample comprising hematopoietic stem cells (HSCs), and ii) a device configured to collect desired cells from the first cell sample by identifying cells according to their positive or negative expression of cell surface proteins; and b) using the device to identify the desired cells from the first cell sample in order to generate a second cell sample, wherein the using the device involves assessing the presence or absence of at least one of the following proteins on the surface of cells within the first cell sample: CD150, CD48, or CD244, wherein the desired cells are HSCs, and wherein the desired cells are selected from at least one of the following: CD150+ cells, CD48- cells, or CD244- cells, and c) collecting in the second cell sample HSCs and other cells that are at least one of the following: CD150+, CD48-, or CD244-, wherein the second cell sample comprises a higher percent of HSCs than are present in the first cell sample.
  • HSCs
  • the present invention provides compositions comprising an enriched cell sample, wherein the enriched cell sample comprises HSCs, and wherein at least 55% of the cells in the enriched cell sample are CD150+ HSCs. In further embodiments, at least 60%, or 70%, 80%, or 90% or 95% of the cells in the enriched cell sample are CD150+ HSCs.
  • the present invention provides an enriched cell sample, wherein the enriched cell sample comprises HSCs, and wherein at least 20% of the cells in the enriched cell sample are CD150+CD48- HSCs. In particular embodiments, at least 50%, 75%, or 90% of the cells in the enriched cell sample are CD150+CD48- HSCs.
  • the present invention relates to methods comprising: a) providing; i) a subject, and ii) a composition selected from the group consisting of: A) a first enriched cell sample comprising HSCs, wherein at least 60% of the cells in the first enriched sample are CD150+ HSCs; and B) a second enriched cell sample comprising cells, wherein at least 35% of the cells, when intravenously injected into the subject, are able to home to bone marrow and engraft; and b) treating the subject with the first enriched cell sample or the second enriched cell sample.
  • the subject has been exposed to high dose chemotherapy.
  • at least 80% of the cells in the first enriched cell sample are CD150+ HSCs.
  • at least 35% of the cells in the second purified cell sample are further able to provide long-term multi-lineage reconstitution.
  • the desired cells are CD150 + cells and the purified cell sample comprises a higher percentage of CD150 + HSCs than are present in the test sample.
  • the desired cells are CD48 - cells and the purified cell sample comprises a higher percentage of CD48 - HSCs than are present in the test sample.
  • the desired cells are CD244 - cells and the purified cell sample comprises a higher percentage of CD244 - HSCs than are present in the test sample.
  • the purified cell sample comprises greater than 1 percent CD150 + HSCs.
  • the desired cells are CD150 + CD48 - cells and the purified cell sample comprises a higher percentage of CD150 + CD48 - HSCs than are present in the test sample.
  • the purified cell sample comprises greater than 0.01 percent of CD150 + CD48 - HSCs.
  • the desired cells are CD150 + CD48 - CD244 - cells and the purified cell sample comprises a higher percentage of CD150 + CD48 - CD244 - HSCs than are present in the test sample.
  • the purified cell sample comprises greater than 0.01. percent of CD 150 + CD48 - CD244 - HSCs.
  • the test sample is from a subject.
  • the present invention provides compositions comprising a purified cell sample, wherein the purified cell sample comprises HSCs, and wherein at least 55% of the HSCs in the purified cell sample are either CD150 + , CD48 - , or CD244 - .
  • at least 60% (or at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%) of the HSCs in the purified cell sample are CD150 + , CD48 - , or CD244 - .
  • the present invention relates to compositions comprising a purified cell sample, wherein the purified cell sample comprises cells, and wherein at least 30% of the cells, when intravenously or otherwise injected into an animal, are able to home to bone marrow and engraft.
  • the at least 30% of the cells may be further able to provide long-term multi-lineage reconstitution.
  • at least 40% of the cells, when intravenously or otherwise injected, are able to home to bone marrow and engraft.
  • the animal has been subject to myeloablative treatment.
  • the at least 30% or 40% of the cells are CD150 + , CD48 - , or CD244 - , or any particular combination thereof.
  • the present invention relates to methods comprising a) providing i) a subject, and ii) a composition selected from the group consisting of: A) a first purified cell sample comprising HSCs, wherein at least 50%, 55%, 60% and higher of the cells in the first purified sample are CD150 + HSCs; and B) a second purified cell sample comprising cells, wherein at least 21%, 25%, 35%, 40% and higher of the cells, when intravenously injected into the subject, are able to home to bone marrow and engraft, b) treating the subject with the first or second purified cell sample.
  • the subject has been exposed to high dose or other chemotherapy.
  • the subject may be suffering from aplastic anemia.
  • the present invention relates to methods comprising a) providing i) a test cell sample from a subject, wherein the test cell sample comprises hematopoietic stem cells (HSCs), ii) a purification device configured to purify HSCs or particular types of MPPs from the test cell sample based the presence or absence of no more than two, or no more than 3, or no more than 4 or 5 (or any combination) of the following SLAM family member proteins: CD150, CD84, CD229 (ly-9), CD244, NTB-A, CS1, CD48, CD58, CD2, 19A and Ly108.3; and b) using the purification device on the test cell sample under conditions such that a purified cell sample is generated, wherein the purified cell sample comprises a higher percent of HSCs or a particular type of MPPs than present in the test cell sample.
  • the test sample is a hematopoietic tissue sample.
  • the present invention relates to methods comprising a) providing i) a test cell sample from a subject, wherein the test cell sample comprises hematopoietic stem cells (HSCs) and ii) a purification device configured to isolate, collect, or purify CD150 + HSCs from the test cell sample; and b) contacting the test cell sample with the purification device under conditions such that a purified cell sample is generated, wherein the purified cell sample comprises a higher percent of CAD150 + HSCs than present in the test cell sample.
  • the test sample can be a hematopoietic tissue sample.
  • the purification device can be further configured to purify CD48 - HSCs from the test sample, and wherein the purified cell sample further comprises a higher percent of CD48 - HSCs than present in the test sample.
  • the test cell sample may comprise whole bone marrow (WBM) cells, or cells from any other hematopoietic tissue sample.
  • the purified cell sample comprises at least 1.0% CD150 + HSCs (e.g. of all the cells in the purified cell sample, at least 1.0% are CD150 + HSCs). In some embodiments, the purified cell sample comprises at least 2.0% or at least 3.0% or at least 4.0% CD150 + HSCs, or between 1% and 10% CD150 + HSCs. In additional embodiments, the purified cell sample comprises greater than 5 percent CD150 + HSCs (e.g. of all the cells in the purified cell sample at least 5.1 percent, about 6.0 percent, about 8.0 percent about 10 or greater, such as 5-15 percent are CD150 + HSCs).
  • the purified cell sample comprises greater than 0.01 percent of CD150 + CD48 - HSCs (e.g. of all the cells in the purified cell sample at least 0.02 percent or 0.05 percent or 0.1 percent or 5 percent are CD150 + CD48 - HSCs). In additional embodiments, the purified cell sample comprises greater than 0.01 percent of CD150 + CD48 - CD244 - HSCs (e.g. of all the cells in the purified cell sample at least 0.02 percent or 0.05 percent or 0.1 percent or 5 percent are CD150 + CD48 - CD244 - HSCs).
  • the purification device may comprise a cytometer or similar device.
  • the purification device may comprise anti-CD150 antibodies, antibody fragments, or other CD150 binding molecules.
  • the purification device may comprise anti-CD48 antibodies, antibody fragments, or other CD48 binding molecules.
  • the purification device may comprise anti-CD244 antibodies, antibody fragments, or other CD244 binding molecules.
  • the subject can be a human or a mouse, rat, dog, cat, pig, cow or horse, other mammal, or bird.
  • the present invention provides compositions comprising a purified cell sample, wherein the purified cell sample comprises greater than 1, 2, 3, 4, 5 or 10 percent CD150 + hematopoietic stem cells (HSCs).
  • the purified cell sample is a whole bone marrow cell sample or other hematopoietic tissue sample.
  • the present invention provides compositions comprising a purified cell sample, wherein the purified cell sample comprises greater than 0.01 percent of CD150 + CD48 - hematopoietic stem cells (HSCs).
  • the purified cell sample is a whole bone marrow cell sample.
  • the present invention provides compositions comprising a purified cell sample, wherein the purified cell sample comprises a higher percentage of CD150 + hematopoietic stem cells (HSCs) than present in an unpurified cell sample taken from a subject.
  • the purified cell sample comprises a higher percentage of CD48 - HSCs than present in the unpurified cell sample taken from the subject.
  • the purified cell sample comprises a higher percentage of CD244 - HSCs than present in the unpurified cell sample taken from the subject.
  • the unpurified cell sample comprises whole bone marrow cells.
  • the present invention relates to methods comprising a) providing i) a subject, and ii) a composition selected from the group consisting of: A) a first purified sample comprising greater than 1, 2, 3, 4 or 5 percent CD150 + hematopoietic stem cells (HSCs); B) a second purified sample comprising greater than 0.01 percent of CD150 + CD48 - hematopoietic stem cells (HSCs); C) a third purified sample comprising a higher percentage of CD150 + (or CD48' or CD244 - ) hematopoietic stem cells (HSCs) than present in an unpurified cell sample taken from the subject; D) a fourth purified sample comprising greater than 0.01 percent of CD150 + CD48 - CD244 - hematopoietic stem cells (HSCs); and b) treating the patient with the first, second, third, or fourth purified cell sample.
  • the subject has been exposed to high dose or other chemotherapy.
  • the present invention relates to devices configured for purifying CD150 + HSCs from a test cell sample.
  • the present invention relates to devices (e.g. cytometers) configured for purifying from a test sample HSCs that are CD150 + , CD48 - , or CD244 - (or any combination thereof).
  • kits comprising; a) reagents for targeting the expression or nonexpression of CD 150, CD48, or CD244 and b) an insert sheet comprising directions for employing the reagents to identifying or collecting HSCs that are CD150 + , CD48 - , or CD244 - (or any combination thereof).
  • the present invention relates to methods comprising a) providing i) reagents for detecting HSCs that are CD150 + , CD48 - , or CD244 - (or any combination thereof), and ii) a cell sample or a subject; and b) contacting the reagents with the cell sample or the subject such that the presence, absence or level of HSCs that are CD150 + , CD48 - , or CD244 - (or any combination thereof) is determined.
  • the present invention provides methods of generating an enriched cell sample comprising: purifying cells from a first sample based on the positive expression of CD150 protein or negative expression of CD48 protein or CD244 protein, in order to generate an enriched cell sample that is enriched 2-fold (or 3-fold, 4-fold, 6-fold, 10-fold or 100-fold) in cells selected from the following: CD150 + cells, CD48 - cells, and CD244 - cells, and wherein said enriched cell sample comprises a higher percent of HSCs than are present in said first sample.
  • the present invention relates to a purification device configured to collect HSCs from a test cell sample by identifying cells according to their positive expression of CD150 or negative expression of CD48 or CD244.
  • the purification device may comprise anti-CD150, anti-CD48, and/or anti-CD244 antibodies or antibody fragments (or other binding molecules).
  • the terms “subject” and “patient” refer to any animal, such as a mammal like a dog, cat, bird, livestock, and preferably a human. Specific examples of “subjects” and “patients” include, but are not limited to, individuals who have been exposed to high dose chemotherapy.
  • treating refers to administering the purified or enriched cell samples of the present invention to a subject, and may include, for example, bone marrow transplants (e.g., of cell samples originally from the subject or from a different subject) or any type of introduction of the cell samples of the present invention into or onto the body of a subject.
  • bone marrow transplants e.g., of cell samples originally from the subject or from a different subject
  • any type of introduction of the cell samples of the present invention into or onto the body of a subject.
  • the present invention is defined in the claims and provides better and more efficient methods of identifying, isolating, or collecting hematopoietic stem cells (HSCs) and better and more easily created compositions containing high concentrations of HSCs. It has been determined that cells from bone marrow and other hematopoietic tissues that do not express the surface markers CD48 or CD244 but do express the surface marker CD150 makeup a highly purified population of HSCs and that using appropriate combinations of the expression patterns of these surface markers allows for the isolation and identification of a highly purified population of HSCs.
  • HSCs hematopoietic stem cells
  • the present invention provides methods of isolating, identifying, or collecting CD150 + HSCs and CD150 + CD48 - HSCs and CD48 - HSCs and CD150 + CD244 - HSCs from within a sample of hematopoietic tissue.
  • the present invention also provides purified hematopoietic tissue samples having an increased level of CD150 + HSCs and CD150 + CD48 - HSCs and CD48' HSCs and CD150 + CD244 - HSCs.
  • the present invention further relates to methods of treating or evaluating the treatment of subjects by making use of such methods and compositions.
  • the methods of the present invention can make use of known separation techniques and bioassays for identification, quantification or physical separation of HSCs from non-HSC cells within a hematopoietic tissue sample.
  • the methods and compositions of the present invention may be employed advantageously in a wide variety of clinical and research applications and settings. For convenience, the detailed description is provided in the following sections below: (I) Hematopietic Stem Cells; (II) Exemplary Sources of Hematopoietic Tissue Samples Containing HSCs; (III) Exemplary HSC Purification Techniques; and (IV) Exemplary Embodiments.
  • HSCs Hematopoietic stem cells
  • B-cells B-cells
  • T-cells myeloid cells
  • platelets red blood cells.
  • HSCs In fetal development HSCs exist in the peripheral blood, the umbilical cord blood vessels, the placenta, the aorto-gonado-mesonephros, the fetal liver and spleen, and, eventually, the developing bone marrow cavities.
  • HSCs reside largely in the bone marrow cavities under physiological conditions.
  • HSCs in adults have also been shown to circulate in very low frequencies through the vasculature existing in both the spleen and skeletal muscle.
  • the circulation of HSCs through the circulatory system may be induced using a mobilization procedure such as cyclophosphamide followed by G-CSF treatment (CY/G-CSF), which expands the number of HSCs in the blood and in spleen tissue.
  • CY/G-CSF G-CSF treatment
  • HSCs are capable of self-renewal and long-term multilineage differentiation
  • other hematopoietic progenitor cells capable of giving rise to cells of mature blood lineages after transplantation or in cultures are not capable of self-renewal and produce mature blood cells only transiently.
  • These other hematopoietic progenitor cells which encompass MPPs, are therefore incapable of sustaining long-term hematopoiesis.
  • HSCs give rise to other HSCs following cell division under certain conditions also give rise to non-self-renewing multipotent progenitors (MPPs) that can be isolated from murine hematopoietic tissue as Thy-1.1loSca-1 + Mac-1loCD4lo (See, Morrison et al., 1994 and Morrison et al. 1997 supra). These cells give rise to myeloid, B and T cells for less than 12 weeks after injection into irradiated recipients.
  • MPPs are not able to give rise to a sufficient number of other MPPs to sustain long-term hematopoiesis and are therefore considered to not be able to undergo self-renewal.
  • Tissue and cell samples containing HSCs can be transplanted into diseased, chemotherapeutically conditioned, or lethally (or sublethally) irradiated recipients rescuing the recipients from death owing to hematopoietic failure or compromise, which can result in overwhelming infection or inadequate erythropoiesis.
  • samples must first be purified to isolate, and obtain artificially high concentrations of, HSCs, for example, by detecting expression of specific cell surface proteins or receptors, cell surface protein markers, or other markers.
  • Highly purified HSCs are increasingly being used clinically, in a variety of applications, such as for autologous transplants into patients after high-dose chemotherapy.
  • HSCs with the maximum degree of purity so as to minimize contamination by immune effector cells (such as lymphocytes) or cancer cells. Since few markers have been identified that are highly specific to HSCs, however, it has previously not been possible to achieve the required degree of purity based on simple combinations of one or two markers associated with cell surface proteins. As a result, it has been possible to highly purify or identify HSCs only by making use of complex combinations of many markers.
  • hematopoietic tissue samples for use in various embodiments of the present invention are not limited by source or origin. Such samples can be taken from a subject to be treated or from another individual.
  • Hematopoietic tissue for the identification or isolation of HSCs may include but is not limited to bone marrow, isolated from sternum, iliac crests, femora or other bone marrow cavities.
  • Other sources include any tissue which contains any amount of HSCs but may have only endogenously low frequencies of HSCs. These tissues may include but are not limited to embryonic yolk sac, fetal liver or spleen, adult spleen or treated or untreated peripheral blood, and umbilical cord blood.
  • Cells may be collected by physically or enzymatically or chemically dissociating cells in single cell suspension such that a majority of cells to be further processed are no longer attached to other cells from within the original hematopoietic tissue sample whether through direct cell-cell interactions or indirectly through extracellular connective tissue.
  • Cells may be processed further in an appropriate isoosmotic salt solution such as phosphate buffered saline (PBS) or Hank's buffered saline solution (HBSS; as described herein) which may or may not contain protein in the form of BSA or serum and which may or may not contain chemicals buffers to maintain physiological pH.
  • PBS phosphate buffered saline
  • HBSS Hank's buffered saline solution
  • HSCs may be acquired from primary hematopoietic tissue or alternatively may be isolated or identified prior to culture under conditions conducive to the expansion of hematopoietic progenitors such as by culturing in hematopoietic bioreactors or by supplementation with SCF or by co-culturing with stromal elements supportive of hematopoietic progenitor cell expansion or survival or by culturing in media conditioned by being previously inoculated onto stromal feeder layers which secrete factors and proteins conducive to the maintenance and survival and expansion of HSCs.
  • hematopoietic progenitors such as by culturing in hematopoietic bioreactors or by supplementation with SCF or by co-culturing with stromal elements supportive of hematopoietic progenitor cell expansion or survival or by culturing in media conditioned by being previously inoculated onto stromal feeder layers which secrete factors and proteins conducive to the maintenance and survival and expansion of HSCs
  • hematopoietic tissue samples appropriate for use according to the present invention include tissue samples that have been pre-sorted for HSCs (or MPPs) according to known or other sorting techniques such as sorting for CD34 expression or pre-enriching by density elutriation among other things.
  • the present invention is not limited by the purification technique or device that takes advantage of the presence or absence of SLAM family member proteins (e.g. CD150, CD48, CD244, or any combination thereof) on HSCs, MPPs, or other hematopoietic cells.
  • SLAM family member proteins e.g. CD150, CD48, CD244, or any combination thereof
  • Any method suitable for identifying surface proteins, whether known or to be discovered, could be employed in the various methods of the present invention.
  • HSCs according to the present invention may be identified using fluorescence activated cell sorting analysis (FACS) which typically uses antibodies conjugated to fluorochromes to directly or indirectly assess the level of expression of a given surface protein on individual cells within a heterogenous (or homogenous) cell preparation of hematopoietic tissue.
  • FACS fluorescence activated cell sorting analysis
  • SLAM family member surface proteins in particular, CD150, CD48 and CD244 on individual cells within the cell preparation
  • HSCs may be physically separated from other cells within a cellular preparation of hematopoietic tissue using any previously developed or as yet undeveloped technique whereby cells are directly or indirectly differentiated according to their expression or lack of expression of SLAM- family member surface proteins CD150, CD48, CD244, as well as CD84, CD229, NTB-A, CS1, CD58, CD2, 19A or Ly108.3.
  • Positive selection or negative selection may be employed to enrich for or deplete of, respectively, cells which do or do not express the SLAM-family member cells surface markers (e.g. CD150, CD48, or CD244).
  • Common methods used to physically separate specific cells from within a heterogenous population of cells within a hematopoietic cell preparation include but are not limited to flow-cytometry using cytometer which may have varying degrees of complexity and or detection specifications, magnetic separation, using antibody or protein coated beads, affinity chromatography, or solid-support affinity separation where cells are retained on a substrate according to their expression or lack of expression of a specific protein or type of protein.
  • Such separation techniques need not, but may, completely purify or nearly completely purify (e.g. 99.9% are perfectly separated) HSCs or populations enriched in HSCs according to expression or lack of expression of SLAM family members.
  • the methods of the invention are related to methods that involve the steps of obtaining a hematopoietic tissue sample containing HSCs and/or MPPs, identifying desired cells such as HSCs or particular MPPs from within the sample based on their expression or lack of expression of particular SLAM family member proteins.
  • the methods involve identifying those cells from within a sample that express CD150, do not express CD48, and do not express CD244 (or any combination of these expression characteristics), isolating or collecting the identified cells for quantification, further examination and study, or for use in treatment of a wide variety of medical conditions.
  • the methods and compositions of the present invention can be employed advantageous in a wide variety of applications.
  • the methods and compositions of the present invention can be used.
  • purified HSCs as described in this invention would be useful in bone marrow transplantation as well as mobilized peripheral blood or umbilical cord blood transplantation as well as the transplantation of other organs in association with the transplantation of bone marrow; for the prospective identification and quantification of HSCs resident in unfractionated bone marrow or other hematopoietic tissue transplants or to monitor the progress of a patient's hematopoietic tissue transplantation or chemotherapeutic treatment; for the purposes of studying the properties of HSCs including their response to various growth factors, their production of growth factors, their interaction with stromal elements, etc; for the introduction, amplification and/or modification of endogenous or exogenous genes or gene elements to promote the health of recipients of transplanted HSCs; for the treatment of leukemias or lymphomas
  • HSCs according to the methods and compositions of the present invention would also find use 1) in regenerating the hematopoietic system of a host deficient in HSCs, 2) in a host that is diseased and can be treated by removal of bone marrow or other hematopoietic tissue containing HSCs, isolation of HSCs and treatment of individuals with pharmaceuticals or irradiation prior to re-infusion of HSCs, 3) producing various hematopoietic cells in vivo or in vitro, 4) the development of hematopoietic cell lineages and assaying for factors or biological properties including gene expression profiles associated with hematopoietic development, and 5) treatment of genetic diseases through replacement, amplification, modification or inhibition of genes or gene elements, for example.
  • the identification of HSCs, with or without later transplantation or reconstitution using the identified HSCs can be useful in itself.
  • Identification of HSCs within a hematopoietic tissue sample or cell preparation derived from a hematopoietic tissue sample may be useful for assessing the ability of a sample of hematopoietic cells to give rise to engraftment and reconstitution in patients undergoing bone marrow transplantation; for monitoring the progress of recovery from myeloablative treatment or other therapy damaging to hematopoietic tissues such as irradiation or chemothereutic protocols; for assessing the HSC content of hematopoietic cultures; for monitoring the progress, assessing the status of or ascertaining the prognosis for the disease of a patient having a disorder of the hematopoietic system such as aplastic anemia or lymphoma or a neoplastic disorder such as leukemia or diseases conferring immunocompromise; and for treatment of intractable disorders associated with
  • HSCs may be used for the treatment of genetic diseases or for the treatment of diseases which may be ameliorated by modification of gene expression within a host.
  • Genes or gene elements may be introduced, inhibited, removed, modified, or otherwise altered in the HSCs of patients undergoing autologous or allogeneic HSC or hematopoietic cell transplant.
  • HSCs may be useful for treatment of genetic diseases associated with the hematopoietic system such as the red blood cell dyscrasias, beta-thalassemia, sickle cell anemia, adenosine deaminase deficiency or diseases associated with clotting factor deficiency such as hemophilia or diseases associated with immunocompromise such as recombinase deficiency or severe-combined immuno-deficiency syndrome. Genetic manipulation of HSCs may also be useful for treatment of non-hematopoietic disorders where the disease is associated with the absence of an appropriately expressed or appropriately secreted protein product such as a hormone or enzyme.
  • genetic diseases associated with the hematopoietic system such as the red blood cell dyscrasias, beta-thalassemia, sickle cell anemia, adenosine deaminase deficiency or diseases associated with clotting factor deficiency such as hemophilia or diseases associated with immunocompromise such as recombina
  • Genes or gene elements may also be introduced into HSCs by using the ability of self-ligating properties of CD150 (or other proteins) across the membranes of two separate cells expressing CD150 or the self-liganding properties of CD150 within the membrane of an individual cell expressing CD150 protein.
  • Treatment of hematopoietic tissue including HSCs with certain clones ofanti-CD150 antibodies may be used to activate or modify gene expression or augment or alter the biological activity of HSCs or may disrupt the interaction of CD 150 on the surface of HSCs and on the surface of other cellular elements within hematopoietic tissue or hematopoietic stromal co-culture to modulate the ability of said co-culture to maintain or expand numbers of HSCs within said co-culture.
  • Treatment of hematopoietic tissue or HSCs with certain clones of anti-CD150 antibodies may also be used disrupt or augment the ability of HSCs to home to and engraft in hematopoietic microenvironments suitable for the maintenance and expansion of HSCs.
  • MPPs capable of only transient reconstitution in vivo and only transient hematopoietic cell production in vitro may be purified according to their expression of SLAM-family members.
  • MPPs may be identified or isolated as cells which do not express CD150, or which do not express CD48 or which do or do not express CD244.
  • Isolation or identification of MPPs from within a heterogenous cell preparation from a hematopoietic sample may be useful when only transient production of blood cells is desired or when more rapid reconstititution is beneficial to the recipients; for assessing the ability of a sample of hematopoietic cells to give rise to engraftment and reconstitution in patients undergoing bone marrow transplantation; for monitoring the progress of recovery from myeloablative treatment or other therapy damaging to hematopoietic tissues such as irradiation or chemotherapeutic protocols; and for assessing the MPP content of hematopoietic cultures as described previously for HSCs; for monitoring the progress, assessing the status of or ascertaining the prognosis for the disease of a patient having a disorder of the hematopoietic system, among other things; for the transplant of MPPs into subjects alone or in combination with prior or subsequent transplantation of HSCs.
  • the present invention may also be employed to isolate and identify mesenchymal and nervous system progenitors.
  • Progenitors from tissues other than hematopoietic tissue may be isolable or identifiable by their expression pattern of SLAM family members including but not limited to CD150, CD48 and or CD244.
  • the sub-ventricular zone (sVZ) and the developing fetal forebrain (FB) have been shown to contain cells capable of generating large numbers of neurons and glial-lineage cells within the central nervous system.
  • a certain proportion of cells resident within the sVZ of adult animals and within the FB of fetal animals express the SLAM family members CD150; certain of these cells expressing CD150 also express CD48 whereas certain others of these cells do not express CD48.
  • stromal cells resident within the bone marrow including but not limited to cells of the osteoblastic lineage which are the precursors to mature bone cell osteocytes and have been shown to support the maintenance and expansion of HSC numbers may be isolable or identifiable according to their expression of CD 150.
  • Osteoblastic cells within adult bone marrow have been shown to express N-cadherin but not the hematopoietic surface marker CD45.
  • a population of cells exists which expresses N-cadherin, which does not express CD45 and which does express CD150 exists within the adult bone marrow cavity and may be demonstrated to be useful for production of stromal cells supportive of HSCs maintenance or expansion in vitro or for the production of osteocytes or graftable bone tissue precursors.
  • HSCs hematopoietic stem cells
  • MPPs multipotent progenitors
  • Thy-1 low Sca-1 + Lineage - c- kit + cells are highly enriched for long-term reconstituting, multipotent HSCs.
  • Cells injected Mice that engrafted Frequency of cells that engrafted Engrafted mice with long-term multilineage reconstitution Frequency of cells that long-term multilineage reconstituted (HSCs) 10 24/28 1 in 5.7 88% (21/24) 1 in 7.7 (21/28) 5 13/15 1 in 3.0 100% (13/13) 1 in 3.0 (13/15) 4 25/34 1 in 3.5 92% (23/25) 1 in 4.1 (23/34)
  • the indicated number of donor-type (CD45.1 + ) Thy-1 low Sca-1 + Lineage - c-kit + cells were transplanted intravenously into lethally irradiated recipients (CD45.2 + ) along with 200,000 recipient-type (CD45.2 + ).
  • Recipients were considered engrafted by donor cells if any CD45.1 + cells were detected in their peripheral blood (above background: >0.1-0.3% of myeloid cells or >0.1-0.15% of lymphoid cells, depending on the experiment) at least 4 weeks after reconstitution.
  • the frequency of cells that engrafted was calculated based on limit-dilution (Poisson) statistics. Mice were considered long-term multilineage reconstituted if donor-type myeloid, B, and T cells were present for more than 16 weeks after reconstitution.
  • Thy-1 low Sca-1 + Lineage - c- kit + HSCs give rise to non-self-renewing MPPs in vivo (Morrison et al. 1997, supra), prior to undergoing lineage restriction. These MPPs are highly enriched in the Thy-1 low Sca-1 + Mac-1 low CD4 low population. In previous studies, only 26% of Thy-1 low Sca-1 + Mac-1 low CD4 low cells formed myeloerythroid colonies in methylcellulose cultures, and many clones gave rise to only B lineage reconstitution in vivo (Morrison et al. 1994 and Morrison et al. 1997).
  • Thy-1 low Sca-1 + Mac-1 low CD4 low B220 - cells were injected into each of 16 irradiated recipients, 1 out of every 4.0 i.v. injected cells detectably reconstituted.
  • 11 recipients that were reconstituted with donor cells 64% were transiently multilineage reconstituted (M+B+T for ⁇ 8 weeks), 27% were reconstituted only in the B and T lineages, and 9% were reconstituted only in the B lineage.
  • Thy-1 low Sca-1 + Mac-1 low CD4 low cells reconstitute transiently and lack detectable self-renewal potential (Morrison et al., 1997). This indicates that Thy-1 low Sca-1 + Mac-1 low CD4 low B220 - cells are more highly enriched for MPPs than any previously characterized cell population.
  • 1151 probe sets were identified that satisfied these criteria for being upregulated in HSCs as compared to CD45 + cells, and 46 probe sets against unique genes in HSCs as compared to MPPs (out of 36,701 probe sets total). Twenty-seven of these 46 probe sets were expressed at higher levels in HSCs as compared to both MPPs and CD45 + cells. To further evaluate these candidates, their expression was compared by quantitative (real-time) RT-PCR in at least two independent samples of HSCs, MPPs, and whole bone marrow cells. Of the 25 genes against which qPCR primers could be designed, all were confirmed as being expressed at >1.9-fold higher levels in HSCs as compared to MPPs and CD45 + cells (Table 2).
  • Table 2 Genes that were expressed at higher levels in HSCs as compared to MPPs and CD45 + cells by both Microarray analysis and quantitative PCR.
  • the average untransformed probe intensities from three independent samples were used to calculate fold-change (HSC/MPP; HSC/CD45 + ).
  • the table lists all of the genes that were expressed at significantly higher levels in HSCs as compared to MPPs and CD45 + cells by both microarray analysis (fold change>3) and quantitative (real-time) PCR (fold change> 1.9).
  • CD150 One of the few genes that was strongly upregulated in HSCs as compared to MPPs and CD45 + cells, CD150, was particularly interesting because it was not identified as being expressed or upregulated in stem cells in any previously published studies of which the inventors are aware.
  • CD150 appeared to be 4-fold upregulated in HSCs as compared to MPPs and CD45 + bone marrow cells by microarray analysis (Table 2), though this is likely an underestimate of the difference in expression since CD150 was not detectable in MPPs or CD45 + cells by microarray (therefore these samples were set to 100 for the purpose of calculating fold-change (Iwashita et al., 2003).
  • CD150 was upregulated in HSCs by 5.8 ⁇ 1.8-fold as compared to MPPs and 17.0 ⁇ 4.0-fold as compared to whole bone marrow cells (Table 2; Fig 1 ).
  • CD150 was differentially expressed among HSCs and MPPs at the protein level. Only 6.6 ⁇ 1.7% of whole bone marrow cells were CD150 + ( Figure 1A ). Consistent with the trends observed at the RNA level (Table 2), CD150 was expressed by 46 ⁇ 12% of cells within the Thy-1 low Sca-1 + Lineage - c-kit + HSC population but by only 0.9 ⁇ 0.5% of cells in the Thy-1 low Sca-1 + Mac-1 low CD4 low B220 - MPP population ( Figure 1B,C ).
  • CD150 + cells included HSCs
  • 20,000 donor-type CD150 + cells or 180,000 donor-type CD150 - bone marrow cells cell doses were based on the fraction of 200,000 whole bone marrow cells that were CD150 + or CD150 - as in prior HSC marker studies (Uchida et al., supra) transplanted into lethally irradiated recipients along with a radioprotective dose of 200,000 recipient-type whole bone marrow cells ( Fig. 1D ).
  • recipients of the CD150 + cells were always long-term multilineage reconstituted by donor cells (6/6 recipients) while recipients of the CD150 - cells usually (8/9 recipients) exhibited transient multilineage reconstitution.
  • recipients of the CD150 - cells usually (8/9 recipients) exhibited transient multilineage reconstitution.
  • 30,000 C57BL whole bone marrow cells yield long-term multilineage reconstitution in around 60% of recipients ( Uchida et al.., J Exp Med. 1992 Jan 1;175(1):175-84 )
  • HSCs are enriched in the CD150 + fraction and depleted in the CD150 - fraction of bone marrow cells.
  • Recipients of CD150 + whole bone marrow cells were consistently able to transfer long-term multilineage donor cell reconstitution to secondary recipients, while recipients of CD150 - cells were not able to transfer donor cell reconstitution to secondary recipients (data not shown).
  • SLAM family member CD244 which was not detected in HSCs by microarray analysis, was examined. At the protein level, only 8.9 ⁇ 0.6% of bone marrow cells expressed CD244 by flow-cytometry ( Fig. 2A ). Little or no CD244 staining was detected among Thy-1 low Sca-1 + Lineage - c-kit + HSCs ( Fig. 2B ) but 33 ⁇ 10% of cells in the Thy-1 low Sca-1 + Mac-1 low CD4 low MPP population were CD244 + ( Fig. 2C ).
  • CD244 + cells were depleted of HSC activity while retaining transiently reconstituting progenitors
  • two independent competitive reconstitution assays were performed in which 20,000 donor-type CD244 + cells or 180,000 donor-type CD244 - bone marrow cells were transplanted into lethally irradiated recipients along with a radioprotective dose of 200,000 recipient-type whole bone marrow cells ( Fig. 2D ).
  • Recipients of the CD244 - cells were consistently long-term multilineage reconstituted by donor cells (8/8 recipients) while recipients of the CD244 + cells were consistently transiently multilineage reconstituted (8/8 recipients).
  • the SLAM family member CD48 was expressed at significantly higher levels on CD45 + cells as compared to HSCs or MPPs (fold change>3).
  • CD48 protein expression was also reduced on HSCs: 43 ⁇ 3% of whole bone marrow cells were CD48 + ( Fig. 3A ) while only 23 ⁇ 9% of Thy-1 low Sca-1 + Lineage - c-kit + cells were CD48 + ( Fig. 3B ).
  • CD48 + cells were depleted of HSC activity.
  • Recipients of the CD48 - cells were consistently long-term multilineage reconstituted by donor cells (5/5 recipients) while recipients of the CD48 + cells were consistently reconstituted by B cells (6/6 recipients), but never by myeloid cells (0/6) and rarely by T cells (1/6).
  • HSCs are enriched in the CD48 - fraction and depleted in the CD48 + fraction of bone marrow cells.
  • CD48 was not expressed on multipotent progenitors, it was expressed by the majority of progenitors that formed myeloerythroid colonies in culture (data not shown) in addition to early B lineage progenitors ( Fig. 3D ).
  • CD 150 was preferentially expressed by HSCs
  • CD48 was preferentially expressed by restricted hematopoietic progenitors.
  • mice were competitively reconstituted with single CD150 + CD48-Sca-1 + Lineage-c-kit + cells in five independent experiments (Table 4). Only 0.003 ⁇ 0.0009% of bone marrow cells were CD150 + CD48 - Sca-1 + Lineage - c-kit + .
  • One CD150 + CD48-Sca-1 + Lineage - c-kit + cell was sorted per well and then the contents of each well were individually injected into the recipients. Visual inspection confirmed the presence of a single cell per well prior to injection, and functionally confirmed the presence of a single cell per well in control studies ( Figure 5 ).
  • mice Of the 10 recipients injected with CD150 - Thy-1 low Sca-1 + Lineage - c-kit + cells, most mice showed no donor cell reconstitution. This demonstrates that the combination of markers for CD150 and CD48 with previously identified HSC markers significantly increases HSC purity.
  • Table 4 Competitive reconstitution of irradiated mice with single CD150 + CD48 - Sca-1 + Lineage c-kit + cells revealed that these cells were highly enriched for long-term reconstituting multipotent HSCs.
  • Hematopoietic stem cell populations (Mac-1 - CD4 - c-kit + ; Mac-1 lo CD4 lo ) for identification of CD150 and other SLAM family member expression profiles on HSCs and hematopoietic precursors were isolated as previously described (Morrison et al., 1994 and Morrison et al., 1997, supra). Briefly, whole bone marrow or other hematopoietic tissue cells were incubated with unconjugated monoclonal antibodies to lineage specific surface molecules including B220 (6B2), CD3 (KT31.1), CD5 (53-7.3), CD8 (53-6.7), Gr-1 (8C5) and Ter119.
  • B220 6B220
  • CD3 KT31.1
  • CD5 53-7.3
  • CD8 53-6.7
  • Gr-1 (8C5) Gr-1
  • pelleted cells were resuspended in anti-rat IgG specific F(ab) 2 fragment conjugated to phycoerythrin (PE; Jackson ImmunoResearch, West Grove PA). Cells were subsequently stained with directly conjugated antibodies to Sca-1 (Ly6A/E; allophycocyanin (APC)), c-kit (2B8; biotin), Thy-1.1 (19XE5; fluorescein-5-isothiocyanate (FITC)), Mac-1 (M1/70; PE) and CD4 (GK1.5; PE).
  • CD150 Cells sorted according to expression of CD150 were incubated with unconjugated antibody to CD150 (26D12 received from DNAX, Palo Alto CA), and subsequently stained with goat anti-rat IgG specific F(ab) 2 fragment conjugated to FITC (Jackson ImmunoResearch) or goat anti-rat IgG (H+L) APC (Jackson ImmunoResearch). Cells sorted according to CD48 expression were stained with directly conjugated CD48 (FITC or PE; Pharmingen, San Jose, CA).
  • Methylcellulose culture was performed as previously described (Morrison et al., 1994). Briefly, unfractionated bone marrow cells, unfractionated splenocytes, or single resorted hematopoietic progenitors were plated in the wells of a 96-well plate (Coming, Corning NY) containing 100 ⁇ l 1.0% methylcellulose (Stem Cell Technologies, Vancouver BC).
  • the methylcellulose was supplemented with 20% charcoal absorbed fetal bovine serum (Cocalico, Reamstown PA), 1% BSA (Sigma), 1% penicillin/streptomycin (Gibco) 50ng/ml stem cell factor (SCF), 10ng/ml interleukin-3 (IL-3), 10ng/ml interleukin-6 (IL-6), 3U/ml erythropoietin (Epo), 10ng/ml Flt-3 and 10ng/ml thrombopoietin (Tpo). All cytokines for hematopoietic culture were obtained from R&D Systems (Minneapolis MN). Colonies were maintained at 37°C in fully humidified chambers containing 6% CO 2 . Colony formation was scored after 10-14 days of culture.
  • CD45.1 + stem or progenitor cells were resorted into individual wells of a 96-well plate containing 2-3x10 5 CD45.2 + whole bone marrow cells in 100 ⁇ l HBSS + .
  • peripheral blood was obtained from the tail veins of individual recipients, subject to ammonium-chloride red cell lysis, and stained with antibodies to CD45.1 (104) directly conjugated to FITC, and B220 (6B2), Mac-1 (M1/70), CD3 (KT31.1) and Gr-1 (8C5) directly conjugated to PE or APC as described (Morrison et al., 1994 and Morrison et al., 1997).
  • first strand was synthesized using T7-d(T) 24 primer.
  • complementary RNA was generated by T7 RNA polymerase (Promega, Madison WI).
  • first strand cDNA was synthesized using random hexamers and second strand was synthesized using the T7-d(T) 24 primer.
  • the double stranded cDNA was resuspended with 22 ⁇ l RNase-free water and transcribed to cRNA with the biotin labeling kit (BioArray Highyield RNA transcript labeling kit (T7), Enzo Diagnostics, Farmingdale NY) for twelve hours.
  • cRNA was purified using the RNeasy Mini Kit. Around 60 ⁇ g of biotinylated cRNA were obtained from two rounds of RNA amplification from 5,000 HSCs.
  • HSC cRNA After fragmentation, 15 ⁇ g of HSC cRNA were hybridized per chip to Mouse Genome U74 Arrays (Chips A, B and C; AFFYMETRIX). The chips were hybridized and scanned according to the manufacturer's instructions. Signal intensities were read and analyzed using methods described previously ( Iwashita et al., Science 2003; 301:972 , herein incorporated by reference). To measure fold changes, all negative signal intensity values or values less than 100 were set to 100. To calculate the squared Pearson's correlation coefficient (R 2 ) between two groups, we transformed each value to the base 10 logarithm (log 10 ). Log 10 transformation is required because the Pearson's correlation coefficient is designed to be calculated based on normally distributed data, and the untransformed data are not normally distributed. The statistical significance of differences in signal intensity for each probe set were evaluated by student's T-test using the log 10 transformed values from 3 independent replicates per cell type.
  • RNA was then purified using an RNeasy Mini Kit (Qiagen, Valencia CA) according to manufacturer's instructions and washed three times with 500 ⁇ l RNase-free water.
  • the RNA was used for making cDNA by reverse transcription with 1 ⁇ g random hexamer.
  • the cDNA was extracted with phenolchloroform and precipitated with 20 ⁇ g glycogen. After dissolving the cDNA with RNase-free water, cDNA equivalent to 200 cells was used for each PCR reaction. qRT-PCR was performed in triplicate using three independent cell samples. Primers were designed to have a Tm of ⁇ 59°C and to generate short amplicons (100-150bp).
  • RNA content of samples compared by qRT-PCR was normalized based on the amplification of hypozanthine phosphribosyl transferase (HPRT).
  • HPRT hypozanthine phosphribosyl transferase
  • qRT-PCR products were separated in 2% agarose gels to confirm the presence of a single band of the expected size.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Claims (21)

  1. Procédé comprenant: la purification de cellules à partir d'un premier échantillon cellulaire, basé sur i) l'expression positive de la protéine CD150, ou ii) l'expression négative de la protéine CD48, ou iii) l'expression négative de la protéine CD244, afin de générer un échantillon cellulaire purifié, où lesdites cellules sont sélectionnées parmi au moins un des éléments suivants : des cellules CD150+, des cellules CD48- et des cellules CD244-, et où ledit échantillon cellulaire purifié comprend un pourcentage de CSH plus élevé que celui qui est présent dans ledit premier échantillon cellulaire.
  2. Procédé selon la revendication 1, dans lequel lesdites cellules sont des cellules CD48-, et dans lequel ledit échantillon cellulaire purifié comprend un pourcentage de CSH CD48- plus élevé que celui qui est présent dans ledit premier échantillon cellulaire.
  3. Procédé selon la revendication 1, dans lequel lesdites cellules sont des cellules CD150+CD244-, et dans lequel ledit échantillon cellulaire purifié comprend un pourcentage de CSH CD150+CD244- plus élevé que celui qui est présent dans ledit premier échantillon cellulaire.
  4. Procédé selon la revendication 1, dans lequel ledit échantillon cellulaire purifié comprend plus de 1 pour cent de CSH CD150+.
  5. Procédé selon la revendication 1, dans lequel ledit échantillon cellulaire purifié comprend plus de 20 pour cent de CSH CD150+CD48-.
  6. Procédé selon la revendication 1, dans lequel lesdites cellules sont des cellules CD150+CD48-, et dans lequel ledit échantillon cellulaire purifié comprend un pourcentage de CSH CD150+CD48- plus élevé que celui qui est présent dans ledit premier échantillon cellulaire.
  7. Procédé selon la revendication 6, dans lequel ledit échantillon cellulaire purifié comprend plus de 0,01 pour cent de CSH CD150+CD48-.
  8. Procédé selon la revendication 1, dans lequel lesdites cellules sont des cellules CD244-CD150+CD48-, et dans lequel ledit échantillon cellulaire purifié comprend un pourcentage de CSH CD244-CD150+CD48- plus élevé que celui qui est présent dans ledit premier échantillon cellulaire.
  9. Procédé selon la revendication 8, dans lequel ledit échantillon cellulaire purifié comprend plus de 0,01 pour cent de CSH CD244-CD150+CD48-.
  10. Procédé selon la revendication 1, dans lequel ledit premier échantillon cellulaire est prélevé chez un sujet.
  11. Composition comprenant un échantillon cellulaire enrichi, dans laquelle ledit échantillon cellulaire enrichi comprend des CSH, et dans laquelle au moins 0,01 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-.
  12. Composition selon la revendication 11, dans laquelle au moins 0,05 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-.
  13. Composition selon la revendication 11, dans laquelle au moins 0,1 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-.
  14. Composition selon la revendication 11, dans laquelle au moins 5,0 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-.
  15. Composition comprenant un échantillon cellulaire enrichi, dans laquelle ledit échantillon cellulaire enrichi comprend des CSH, et dans laquelle au moins 0,01 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-CD244-.
  16. Composition selon la revendication 15, dans laquelle au moins 0,02 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-CD244-.
  17. Composition selon la revendication 15, dans laquelle au moins 0,05 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-CD244-.
  18. Composition selon la revendication 15, dans laquelle au moins 0,1 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-CD244-.
  19. Composition selon la revendication 15, dans laquelle au moins 5,0 % desdites cellules dans ledit échantillon cellulaire enrichi sont des CSH CD150+CD48-CD244-.
  20. Composition selon la revendication 11, dans laquelle ledit au moins 0,01 % est au moins 20 %.
  21. Composition selon la revendication 11, dans laquelle ledit au moins 0,01 % est au moins 50 %.
EP20040809807 2003-09-26 2004-09-27 Identification et isolation de cellules souches hematopoietiques Active EP1687411B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US50611103P 2003-09-26 2003-09-26
PCT/US2004/031677 WO2005030040A2 (fr) 2003-09-26 2004-09-27 Identification et isolation de cellules souches hematopoietiques

Publications (3)

Publication Number Publication Date
EP1687411A2 EP1687411A2 (fr) 2006-08-09
EP1687411A4 EP1687411A4 (fr) 2007-07-11
EP1687411B1 true EP1687411B1 (fr) 2015-05-06

Family

ID=35134711

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20040809807 Active EP1687411B1 (fr) 2003-09-26 2004-09-27 Identification et isolation de cellules souches hematopoietiques

Country Status (3)

Country Link
US (3) US7510877B2 (fr)
EP (1) EP1687411B1 (fr)
WO (1) WO2005030040A2 (fr)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6599710B1 (en) 1999-03-10 2003-07-29 The General Hospital Corporation Treatment of autoimmune disease
US7628988B2 (en) 2002-06-27 2009-12-08 The General Hospital Corporation Methods and compositions for treating type 1 diabetes
US7582313B2 (en) * 2002-06-27 2009-09-01 The General Hospital Corporation Methods of organ regeneration using Hox 11-expressing pluripotent cells
US20070231306A1 (en) * 2005-02-24 2007-10-04 The Scripps Research Institute Isolated myeloid-like cell populations and methods of treatment therewith
US20080025953A1 (en) * 2006-07-25 2008-01-31 Kiminobu Sugaya Vigor Enhancement of Animals Via Administration of Stem Cells
JP5646990B2 (ja) 2007-04-23 2014-12-24 ストワーズ インスティテュート フォー メディカル リサーチ 幹細胞自己複製のための方法及び組成物
DK2953634T3 (da) 2013-02-07 2021-08-30 Massachusetts Gen Hospital Fremgangsmåder til udvidelse eller udtømning af regulerende t-celler
EP3191596B1 (fr) 2014-09-11 2019-10-09 The U.S.A. as represented by the Secretary, Department of Health and Human Services Vecteur lentiviral pour le traitement de troubles de l'hémoglobine
CN115043943A (zh) 2015-05-15 2022-09-13 综合医院公司 拮抗性抗肿瘤坏死因子受体超家族抗体
US11266730B2 (en) 2015-09-29 2022-03-08 The General Hospital Corporation Methods of treating and diagnosing disease using biomarkers for BCG therapy
WO2017059241A1 (fr) 2015-10-02 2017-04-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Système d'administration de protéine lentivirale pour l'édition génomique guidée par l'arn
KR102410778B1 (ko) 2016-05-13 2022-06-21 더 제너럴 하스피탈 코포레이션 길항성 항-종양 괴사 인자 수용체 슈퍼패밀리 항체
WO2019050841A1 (fr) * 2017-09-05 2019-03-14 Bluebird Bio, Inc. Composition de cellules souches hématopoïétiques
EP3511412A1 (fr) 2018-01-12 2019-07-17 Genethon Cellules souches hematopoietiques genetiquement modifiees comme plateforme pour expression des proteines
JP7469328B2 (ja) 2019-04-12 2024-04-16 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル 遺伝子操作された造血幹細胞によるベータサラセミア表現型の補正
AU2021338361A1 (en) 2020-09-03 2023-04-06 Chen, Irvin S.Y Soluble alkaline phosphatase constructs and expression vectors including a polynucleotide encoding for soluble alkaline phosphatase constructs
EP4242297A1 (fr) * 2020-11-03 2023-09-13 EdiGene (GuangZhou) Inc. Préparation cellulaire, utilisation de protéine pour la caractérisation de cellules souches hématopoïétiques, et procédé de détermination de cellules souches hématopoïétiques

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5646001A (en) * 1991-03-25 1997-07-08 Immunivest Corporation Affinity-binding separation and release of one or more selected subset of biological entities from a mixed population thereof
US6353150B1 (en) 1991-11-22 2002-03-05 Hsc Research And Development Limited Partnership Chimeric mammals with human hematopoietic cells
JPH06269293A (ja) 1993-03-19 1994-09-27 Sumitomo Electric Ind Ltd マウスcd48に対するモノクローナル抗体
AU685506B2 (en) * 1993-08-25 1998-01-22 Systemix, Inc. Method for producing a highly enriched population of hematopoietic stem cells
US5840502A (en) * 1994-08-31 1998-11-24 Activated Cell Therapy, Inc. Methods for enriching specific cell-types by density gradient centrifugation
JPH1094390A (ja) 1996-09-20 1998-04-14 Tosoh Corp 新規末梢血幹細胞製造法
JPH10136978A (ja) 1996-11-08 1998-05-26 Otsuka Pharmaceut Co Ltd 造血幹細胞の培養方法
AU5448099A (en) * 1998-09-02 2000-03-27 Chugai Seiyaku Kabushiki Kaisha Method for preparing cell fraction containing hematopoietic stem cells

Also Published As

Publication number Publication date
EP1687411A4 (fr) 2007-07-11
EP1687411A2 (fr) 2006-08-09
WO2005030040A2 (fr) 2005-04-07
US20050158857A1 (en) 2005-07-21
US7919316B2 (en) 2011-04-05
US20090227019A1 (en) 2009-09-10
WO2005030040A8 (fr) 2005-10-20
US8383404B2 (en) 2013-02-26
US7510877B2 (en) 2009-03-31
WO2005030040A3 (fr) 2005-09-15
US20110143430A1 (en) 2011-06-16

Similar Documents

Publication Publication Date Title
US8383404B2 (en) Hematopoietic stem cell identification and isolation
JP2020029458A (ja) 改善された造血幹細胞および前駆細胞療法
KR101445337B1 (ko) 지방 흡인 후의 지방흡인물로부터 조혈모세포의 분리 및 정제
Petit-Cocault et al. Dual role of Mpl receptor during the establishment of definitive hematopoiesis
JP2022172252A (ja) 造血移植片の改善方法
US20160151421A1 (en) Methods and compositions for long term hematopoietic repopulation
US20190046578A1 (en) Methods and compositions for expanding long-term hematopoietic stem cell populations
AU2006217541A1 (en) Method of obtaining a population of human haemopoietic stem cells
JP6838750B2 (ja) Nk細胞を含む細胞集団の製造方法
EP1751661B1 (fr) Procede de croissance, de selection et d'enrichissement selectifs de populations de cellules souches/cellules progenitrices
US20140154219A1 (en) Methods and compositions for large-scale isolation of very small embryonic-like (vsel) stem cells
Mizokami et al. Preferential expansion of human umbilical cord blood-derived CD34-positive cells on major histocompatibility complex-matched amnion-derived mesenchymal stem cells
WO1998012304A1 (fr) Systeme de mise en culture de cellules souches hematopoietiques
JP2004350601A (ja) 霊長類動物の胚性幹細胞から造血系細胞への分化方法
JP2021035400A (ja) Nk細胞を含む細胞集団の製造方法
WO1996015259A2 (fr) Methode de mesure de cellules souches a lignage multiple et son procede d'utilisation
Broxmeyer Use of the cord blood
Sekulovic Characterization of mouse hematopoietic stem cells primed to actively self-renew by NUP98-HOXA10hd fusion gene
WO2009129288A1 (fr) Cellules souches hématopoïétiques caractérisées par l’expression de jam-c

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20060426

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL HR LT LV MK

RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 33/567 20060101ALI20060901BHEP

Ipc: C12N 5/00 20060101AFI20060901BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20070612

17Q First examination report despatched

Effective date: 20080116

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20141219

RIN1 Information on inventor provided before grant (corrected)

Inventor name: KIEL, MARK J.

Inventor name: IWASHITA, TOSHIHIDE

Inventor name: MORRISON, SEAN

Inventor name: YILMAZ, OMER H.

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL HR LT LV MK

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 725724

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150615

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602004047146

Country of ref document: DE

Effective date: 20150618

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 725724

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150506

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20150506

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150907

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150807

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150806

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602004047146

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150506

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20160209

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150927

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20160531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150930

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150930

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20040927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150506

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20180927

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20180927

Year of fee payment: 15

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602004047146

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200401

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20190927

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190927